51
|
Chiloiro S, Giampietro A, Frara S, Bima C, Donfrancesco F, Fleseriu CM, Pontecorvi A, Giustina A, Fleseriu M, De Marinis L, Bianchi A. Effects of Pegvisomant and Pasireotide LAR on Vertebral Fractures in Acromegaly Resistant to First-generation SRLs. J Clin Endocrinol Metab 2020; 105:5588033. [PMID: 31613969 DOI: 10.1210/clinem/dgz054] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/25/2019] [Indexed: 02/13/2023]
Abstract
PURPOSE Osteopathy is an emerging complication of acromegaly. In somatostatin receptor ligands (SRL)-resistant patients, pegvisomant (PegV) and pasireotide LAR (Pasi) are used for acromegaly treatment, but their effect on skeletal health is still not defined. METHODS In a longitudinal retrospective international study, we evaluated incidence of radiological vertebral fractures (VFs) in 55 patients with acromegaly resistant to first-generation SRL. RESULTS At study entry, prevalent VFs occurred in 23 patients (41.8%). Biochemical acromegaly control was reached in 66.7% of patients on PegV and in 66.7% of patients on Pasi. During the follow-up, incident VFs (iVFs) were detected in 16 patients (29.1%). Occurrence of iVFs was associated with prevalent VFs (P = .002), persistence of active acromegaly (P = .01) and higher value of insulin-like growth factor 1 (IGF-1) during follow-up (P = .03). Among patients with active disease at last visit, iVFs occurred less frequently in patients on treatment with Pasi (25%) compared to PegV (77.8% P = .04), independently of the IGF-1 values (P = .90). In patients who reached biochemical control, 22.7% on PegV and 12.5% on Pasi had iVFs (P = .40). Among both treatment groups, the presence of pre-existent VFs was the main determinant for iVFs. CONCLUSION Our data show for the first time that patients with biochemically active disease treated with Pasi had lower risk of iVFs versus those treated with PegV. It also confirms that the presence of pre-existent VFs was the main determinant for iVFs. Additional studies on larger populations and with longer follow-up are needed to confirm our data and disclose the mechanisms underlying our findings.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Giampietro
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Chiara Bima
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federico Donfrancesco
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cara Maya Fleseriu
- University of Pittsburgh, Pittsburgh, PA, US
- Oregon Health and Science University, Portland, OR, US
| | - Alfredo Pontecorvi
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Laura De Marinis
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Bianchi
- Pituitary Unit, Divisione di Endocrinologia, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
52
|
Tulipano G. How treatments with endocrine and metabolic drugs influence pituitary cell function. Endocr Connect 2020; 9:R14-R27. [PMID: 31905162 PMCID: PMC6993271 DOI: 10.1530/ec-19-0482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/01/2020] [Indexed: 12/14/2022]
Abstract
A variety of endocrine and metabolic signals regulate pituitary cell function acting through the hypothalamus-pituitary neuroendocrine axes or directly at the pituitary level. The underlying intracellular transduction mechanisms in pituitary cells are still debated. AMP-activated protein kinase (AMPK) functions as a cellular sensor of low energy stores in all mammalian cells and promotes adaptive changes in response to calorie restriction. It is also regarded as a target for therapy of proliferative disorders. Various hormones and drugs can promote tissue-specific activation or inhibition of AMPK by enhancing or inhibiting AMPK phosphorylation, respectively. This review explores the preclinical studies published in the last decade that investigate the role of AMP-activated protein kinase in the intracellular transduction pathways downstream of endocrine and metabolic signals or drugs affecting pituitary cell function, and its role as a target for drug therapy of pituitary proliferative disorders. The effects of the hypoglycemic agent metformin, which is an indirect AMPK activator, are discussed. The multiple effects of metformin on cell metabolism and cell signalling and ultimately on cell function may be either dependent or independent of AMPK. The in vitro effects of metformin may also help highlighting differences in metabolic requirements between pituitary adenomatous cells and normal cells.
Collapse
|
53
|
Iglesias P, Magallón R, Mitjavila M, Rodríguez Berrocal V, Pian H, Díez JJ. Multimodal therapy in aggressive pituitary tumors. ACTA ACUST UNITED AC 2019; 67:469-485. [PMID: 31740190 DOI: 10.1016/j.endinu.2019.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/19/2019] [Accepted: 08/01/2019] [Indexed: 01/09/2023]
Abstract
The concept of aggressive pituitary tumor (APT) has been precisely defined in recent years. These tumors are characterized by morphological (radiological or histopathological) data of invasion, proliferative activity superior to that of typical adenomas and a clinical behavior characterized by resistance to standard therapies and frequent recurrences. The absence of cerebrospinal or distant metastases differentiates them from the pituitary carcinoma. APTs account for about 10% of all pituitary neoplasm. Proper diagnostic implies participation not only of radiological and hormonal investigation but also a thorough pathological assessment including proliferation markers and immunohistochemistry for hormones and transcription factors. Surgical resection, aiming gross total resection or tumor debulking, is the mainstay initial therapy in most patients. Most patients with APTs need more than one surgical intervention, pituitary radiation, sometimes on more than one occasion, and multiple sequential or combined medical treatments, to finally be doomed to unusual treatments, such as alkylating agents (temozolomide alone or in combination), molecular targeted therapies, or peptide receptor radionuclide therapy. Multimodal therapy, implemented by experts, preferably in specialized centers with high volume caseload, is the only way to improve the prognosis of patients with these uncommon tumors. The research needs in this area are multiple and include a greater knowledge of the molecular biology of these tumors, establishment of protocols for monitoring and sequencing of treatments, development of multicenter studies and international registries.
Collapse
Affiliation(s)
- Pedro Iglesias
- Department of Endocrinology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain.
| | - Rosa Magallón
- Department of Radiation Oncology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Mercedes Mitjavila
- Department of Nuclear Medicine, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | - Héctor Pian
- Department of Pathology, Hospital Universitario, Ramón y Cajal, Madrid, Spain
| | - Juan J Díez
- Department of Endocrinology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
54
|
Alexandraki KI, Papadimitriou E, Mavroeidi V, Kyriakopoulos G, Xydakis A, Papaioannou TG, Kolomodi D, Kaltsas GA, Grossman AB. Role of Receptor Profiling for Personalized Therapy in a Patient with a Growth Hormone-Secreting Macroadenoma Resistant to First-Generation Somatostatin Analogues. J Pers Med 2019; 9:jpm9040048. [PMID: 31731613 PMCID: PMC6963904 DOI: 10.3390/jpm9040048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 11/23/2022] Open
Abstract
Background: Acromegaly is almost always caused by a pituitary adenoma and is associated with high morbidity and mortality when uncontrolled. Trans-sphenoidal removal of the adenoma is the mainstay of therapy, but fails to control the disease in a significant number of patients who require further treatment. Somatostatin analogues (SSAs) as monotherapy or in combination with growth hormone (GH)-receptor antagonists and/or dopamine agonists are used either alone or in combination following surgical failure to achieve disease control. The use of specific biomarkers may help to individualize the therapeutic plan after surgical failure and direct towards a more personalized approach. Methods: We report a 41-year-old man with acromegaly and residual disease after repeated surgery that was resistant to first-generation SSAs. Results: Biochemical and tumor control were achieved following the administration of a second-generation SSA, pasireotide, combined with pegvisomant, both at maximal doses and along with cabergoline. Histology specimens showed a sparsely-granulated GH-immunostaining pituitary adenoma with intense positivity for somatostatin receptors 2 and 5 and low levels of E-cadherin. Conclusion: Personalized medical therapy guided by currently available biomarkers, such as immunohistochemically-characterized receptor profiling or adhesion molecules, resulted in controlled insulin-like growth factor-1 (IGF-1) and GH levels and symptom alleviation following the combination of three drug-classes.
Collapse
Affiliation(s)
- Krystallenia I. Alexandraki
- Endocrine Unit, 1st Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (E.P.); (V.M.); (A.X.); (D.K.); (G.A.K.)
- Correspondence:
| | - Eirini Papadimitriou
- Endocrine Unit, 1st Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (E.P.); (V.M.); (A.X.); (D.K.); (G.A.K.)
| | - Vasiliki Mavroeidi
- Endocrine Unit, 1st Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (E.P.); (V.M.); (A.X.); (D.K.); (G.A.K.)
| | | | - Antonios Xydakis
- Endocrine Unit, 1st Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (E.P.); (V.M.); (A.X.); (D.K.); (G.A.K.)
| | - Theodoros G. Papaioannou
- First Department of Cardiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Denise Kolomodi
- Endocrine Unit, 1st Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (E.P.); (V.M.); (A.X.); (D.K.); (G.A.K.)
| | - Gregory A. Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (E.P.); (V.M.); (A.X.); (D.K.); (G.A.K.)
| | - Ashley B. Grossman
- Centre for Endocrinology, William Harvey Institute, Barts and the London School of Medicine, E1 2AT London, UK;
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX3 7LE Oxford, UK
| |
Collapse
|
55
|
Ozeri O, Cohen ZR, Hadani M, Nass D, Shimon I, Rubinfeld H. Antibody array strategy for human growth factor secretome profiling of GH-secreting adenomas. Pituitary 2019; 22:344-352. [PMID: 30895501 DOI: 10.1007/s11102-019-00955-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSES To test if the antibody array strategy could be utilized to simultaneously detect the secretion of multiple growth factors by human pituitary GH-adenomas and to measure octreotide-induced alterations. METHODS Specimens of human pituitary adenomas were cultured and incubated with or without octreotide for 24 h. Conditional media were analyzed by human growth factor antibody array and VEGF concentrations were measured by ELISA. Media were also analyzed for GH concentrations. p21 expression levels were examined by Western blot of the specimens lysates. RESULTS The antibody arrays successfully identified growth factors secreted by GH-adenomas in vitro. Octreotide treatment induced both elevations and reductions in growth factors secretion. GH response to octreotide was measured, and in this small-sized study resistant and sensitive GH-adenomas presented with no unique secretome pattern of each of the groups. Octreotide-induced VEGF alterations analyzed by the antibody array and by ELISA were not fully matched. CONCLUSIONS This study suggests that the broad proteomic strategy of antibody arrays may be utilized to study the growth factors secretion pattern of GH-adenomas and its regulation by somatostatin analogs or other compounds.
Collapse
Affiliation(s)
- Orly Ozeri
- Institute of Endocrinology and Felsenstein Medical Research Center, Petach Tikva, Israel
- Rabin Medical Center, Petach Tikva, 49100, Israel
- Sackler School of Medicine, Tel Aviv, Israel
- Tel-Aviv University, Tel Aviv, 69978, Israel
| | - Zvi R Cohen
- Sackler School of Medicine, Tel Aviv, Israel
- Tel-Aviv University, Tel Aviv, 69978, Israel
- Department of Neurosurgery, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Moshe Hadani
- Sackler School of Medicine, Tel Aviv, Israel
- Tel-Aviv University, Tel Aviv, 69978, Israel
- Department of Neurosurgery, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Dvora Nass
- Department of Pathology, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Ilan Shimon
- Institute of Endocrinology and Felsenstein Medical Research Center, Petach Tikva, Israel
- Rabin Medical Center, Petach Tikva, 49100, Israel
- Sackler School of Medicine, Tel Aviv, Israel
- Tel-Aviv University, Tel Aviv, 69978, Israel
| | - Hadara Rubinfeld
- Institute of Endocrinology and Felsenstein Medical Research Center, Petach Tikva, Israel.
- Rabin Medical Center, Petach Tikva, 49100, Israel.
- Sackler School of Medicine, Tel Aviv, Israel.
- Tel-Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
56
|
Coopmans EC, Muhammad A, van der Lely AJ, Janssen JAMJL, Neggers SJCMM. How to Position Pasireotide LAR Treatment in Acromegaly. J Clin Endocrinol Metab 2019; 104:1978-1988. [PMID: 30608534 DOI: 10.1210/jc.2018-01979] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/28/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT Pasireotide long-acting release (LAR) is a somatostatin multireceptor ligand, and in the current consensus criteria pasireotide LAR is considered the second-line medical treatment for acromegaly. We present in this article our recommendations to define the position of pasireotide LAR in the treatment of acromegaly and provide recommendations for the management of pasireotide-induced hyperglycemia. EVIDENCE ACQUISITION Our recommendations are based on our experiences with the pasireotide LAR and pegvisomant (PEGV) combination study and the available basic or clinical articles published in peer-reviewed international journals on pasireotide LAR and acromegaly. EVIDENCE SYNTHESIS In accordance with the current consensus criteria, we recommend pasireotide LAR monotherapy as a second-line therapy in young patients who show tumor growth during first-generation somatostatin receptor ligand (SRL) therapy and in patients who show tumor growth during PEGV therapy. In addition, we recommend pasireotide LAR monotherapy in patients with headache not responsive to first-generation SRL therapy and in patients who experience side effects or are intolerant to PEGV monotherapy. In contrast to the current consensus criteria, we recommend considering combination therapy with pasireotide LAR and PEGV as third-line treatment in patients without diabetes at low PEGV dosages (≤80 mg/week) and in patients with tumor growth or symptoms of active acromegaly during first-generation SRL and PEGV combination therapy. With respect to pasireotide-induced hyperglycemia, we recommend a more liberal strategy of blood glucose monitoring during pasireotide treatment. CONCLUSIONS In contrast to the current consensus criteria, we recommend a more reluctant use of pasireotide LAR therapy for the treatment of acromegaly.
Collapse
Affiliation(s)
- Eva C Coopmans
- Department of Internal Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, CB Rotterdam, Netherlands
| | - Ammar Muhammad
- Department of Internal Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, CB Rotterdam, Netherlands
| | - Aart J van der Lely
- Department of Internal Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, CB Rotterdam, Netherlands
| | - Joseph A M J L Janssen
- Department of Internal Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, CB Rotterdam, Netherlands
| | - Sebastian J C M M Neggers
- Department of Internal Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, CB Rotterdam, Netherlands
| |
Collapse
|
57
|
Varlamov EV, McCartney S, Fleseriu M. Functioning Pituitary Adenomas - Current Treatment Options and Emerging Medical Therapies. EUROPEAN ENDOCRINOLOGY 2019; 15:30-40. [PMID: 31244908 PMCID: PMC6587904 DOI: 10.17925/ee.2019.15.1.30] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
Pituitary adenomas are benign tumours comprising approximately 16% of all primary cranial neoplasms. Functioning pituitary adenomas (prolactinomas, somatotroph, corticotroph, thyrotroph and rarely gonadotroph adenomas) cause complex clinical syndromes and require prompt treatment to reduce associated morbidity and mortality. Treatment approaches include transsphenoidal surgery, medical therapy and radiation. Medical therapy is the primary therapy for prolactinomas, and surgery by a skilled neurosurgeon is the first-line approach for other functioning pituitary adenomas. A multimodal treatment is frequently necessary to achieve biochemical and clinical control, especially, when surgery is not curative or when medical therapy fails. Several emerging, novel, medical treatments for acromegaly, Cushing's disease and prolactinomas are in phase II and III clinical trials and may become effective additions to the current drug armamentarium. The availability of various management options will allow an individualised treatment approach based on the unique tumour type, clinical situation and patient preference.
Collapse
|
58
|
Shen SC, Shen CC, Pu TW, Cheng WY. Long-Term Effects of Intracapsular Debulking and Adjuvant Somatostatin Analogs for Growth Hormone-Secreting Pituitary Macroadenoma: 10 Years of Experience in a Single Institute. World Neurosurg 2019; 126:e41-e47. [PMID: 30716503 DOI: 10.1016/j.wneu.2019.01.125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Long-term effects of endoscopic endonasal transsphenoidal intracapsular debulking and adjuvant somatostatin analogs (SSAs) were evaluated in patients with growth hormone- (GH) secreting pituitary macroadenomas. METHODS We retrospectively reviewed the medical records of 45 patients with acromegalic macroadenoma who underwent endonasal endoscopic transsphenoidal intracapsular debulking and received adjuvant SSAs (octreotide) between 2006 and 2015 who had >1 year of follow-up. To evaluate the predictive factors for 1 year and long-term biochemical outcomes, univariate and multivariate analyses were performed. RESULTS Biochemical remission was achieved in 1 year in 20 of the 45 (44.4%) patients, and in 31 of the 45 patients after long-term adjuvant SSA treatment. Tumor control was achieved in 43 of the 45 (93.3%) patients. The univariate analysis showed age (≥55 years), tumor size (diameter ≤1.5 cm), premedication GH levels (≤2.8 ng/mL), premedication insulin-like growth factor 1 levels (≤2-fold of upper limit of normal range), cavernous sinus invasion (Knops grades 2, 3, and 4), and near-total tumor resection were associated with long-term outcomes. The multivariate analysis showed near-total resection was a significant predictor for long-term outcomes (P = 0.019). There were no new pituitary dysfunctions. The observed complications included one case of cerebrospinal fluid leakage and one case of epistaxis requiring intervention. CONCLUSIONS Intracapsular debulking and adjuvant SSAs are a safe and viable treatment for patients with GH secreting pituitary macroadenoma to achieve biochemical remission and tumor control. Although adjuvant SSA treatment enhances residual tumor control, cavernous sinus invasion impedes the remission of endocrine tumors.
Collapse
Affiliation(s)
- Shih-Chieh Shen
- Department of Surgery, Songshan Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chiung-Chyi Shen
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan; Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan
| | - Ta-Wei Pu
- Department of Surgery, Songshan Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Yu Cheng
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan.
| |
Collapse
|
59
|
Ezzat S, Caspar-Bell GM, Chik CL, Denis MC, Domingue MÈ, Imran SA, Johnson MD, Lochnan HA, Grégoire Nyomba BL, Prebtani A, Ridout R, Ramirez JAR, Van Uum S. PREDICTIVE MARKERS FOR POSTSURGICAL MEDICAL MANAGEMENT OF ACROMEGALY: A SYSTEMATIC REVIEW AND CONSENSUS TREATMENT GUIDELINE. Endocr Pract 2019; 25:379-393. [PMID: 30657362 DOI: 10.4158/ep-2018-0500] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: To clarify the selection of medical therapy following transsphenoidal surgery in patients with acromegaly, based on growth hormone (GH)/insulin-like growth factor 1 (IGF-1) response and glucometabolic control. Methods: We carried out a systematic literature review on three of the best studied and most practical predictive markers of the response to somatostatin analogues (SSAs): somatostatin receptor (SSTR) expression, tumor morphologic classification, and T2-weighted magnetic resonance imaging (MRI) signal intensity. Additional analyses focused on glucose metabolism in treated patients. Results: The literature survey confirmed significant associations of all three factors with SSA responsiveness. SSTR expression appears necessary for the SSA response; however, it is not sufficient, as approximately half of SSTR2-positive tumors failed to respond clinically to first-generation SSAs. MRI findings (T2-hypo-intensity) and a densely granulated phenotype also correlate with SSA efficacy, and are advantageous as predictive markers relative to SSTR expression alone. Glucometabolic control declines with SSA monotherapy, whereas GH receptor antagonist (GHRA) monotherapy may restore normoglycemia. Conclusion: We propose a decision tree to guide selection among SSAs, dopamine agonists (DAs), and GHRA for medical treatment of acromegaly in the postsurgical setting. This decision tree employs three validated predictive markers and other clinical considerations, to determine whether SSAs are appropriate first-line medical therapy in the postsurgical setting. DA treatment is favored in patients with modest IGF-1 elevation. GHRA treatment should be considered for patients with T2-hyperintense tumors with a sparsely granulated phenotype and/or low SSTR2 staining, and may also be favored for individuals with diabetes. Prospective analyses are required to test the utility of this therapeutic paradigm. Abbreviations: DA = dopamine agonist; DG = densely granulated; GH = growth hormone; GHRA = growth hormone receptor antagonist; HbA1c = glycated hemoglobin; IGF-1 = insulin-like growth factor-1; MRI = magnetic resonance imaging; SG = sparsely granulated; SSA = somatostatin analogue; SSTR = somatostatin receptor.
Collapse
|
60
|
Malaysian Consensus Statement for the Diagnosis and Management of Acromegaly. J ASEAN Fed Endocr Soc 2019; 34:8-14. [PMID: 33442131 PMCID: PMC7784186 DOI: 10.15605/jafes.034.01.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/14/2019] [Indexed: 12/17/2022] Open
Abstract
In Malaysia, acromegaly is under-recognised with only 10-15% of the expected number of cases from prevalence estimates, having been diagnosed and managed in established endocrine centres with access to multidisciplinary care. This is mainly due to lack of awareness and standardised approach in diagnosing this disease resulting in delay in diagnosis and management with suboptimal treatment outcomes. This first Malaysian consensus statement on the diagnosis and management of acromegaly addresses these issues and is based on current best practices and latest available evidence so as to reduce the disease burden on acromegaly patients managed in the Malaysian healthcare system.
Collapse
|
61
|
Abreu C, Guinto G, Mercado M. Surgical-pharmacological interactions in the treatment of acromegaly. Expert Rev Endocrinol Metab 2019; 14:35-42. [PMID: 30595057 DOI: 10.1080/17446651.2019.1559729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/13/2018] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Acromegaly requires a multimodal treatment approach that includes surgery by an expert pituitary neurosurgeon, pharmacological treatment with one or more of the available drugs and radiation therapy. These treatment alternatives are not mutually exclusive but rather complement each other when properly indicated in the individual patient. In this review, we summarize and analyze the available data concerning the choice of the surgical approach (microscopy vs. endoscopy) and the interactions between medical treatment with somatostatin analogs and pituitary surgery. AREAS COVERED Technical aspects, complications and outcome of transsphenoidal surgery (TSS); Advantages and disadvantages of the microscopic and endoscopic approaches; Safety and efficacy of somatostatin analogs (SSA); Primary pharmacological therapy versus primary TSS; Benefits of the preoperative treatment with SSA; and the effect of surgical tumor debulking in the therapeutic response to SSA. EXPERT COMMENTARY Continuing efforts at improving surgical techniques and at generating more efficacious pharmacological therapies for acromegaly are likely to improve the outcome of these patients. However, an integral approach of the patient aimed not only at achieving biochemical criteria of cure but also at treating the individual comorbidities is mandatory to improve the quality of life of these patients and to reduce their mortality rate.
Collapse
Affiliation(s)
- Coralys Abreu
- a Endocrinology Service , Centro Medico Nacional 20 de Noviembre, ISSSTE , Mexico City , Mexico
| | - Gerardo Guinto
- b Neurological Center , American British Cowdray Medical Center , Mexico City , Mexico
| | - Moisés Mercado
- c Experimental Endocrinology Unit , Hospital de Especialidades, Centro Medico Nacional S. XXI, IMSS , Mexico City , Mexico
| |
Collapse
|
62
|
|
63
|
Günther T, Tulipano G, Dournaud P, Bousquet C, Csaba Z, Kreienkamp HJ, Lupp A, Korbonits M, Castaño JP, Wester HJ, Culler M, Melmed S, Schulz S. International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature. Pharmacol Rev 2018; 70:763-835. [PMID: 30232095 PMCID: PMC6148080 DOI: 10.1124/pr.117.015388] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.
Collapse
Affiliation(s)
- Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Giovanni Tulipano
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Pascal Dournaud
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Corinne Bousquet
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Zsolt Csaba
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Kreienkamp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Márta Korbonits
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Justo P Castaño
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Wester
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Michael Culler
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Shlomo Melmed
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| |
Collapse
|
64
|
Zahr R, Fleseriu M. Updates in Diagnosis and Treatment of Acromegaly. EUROPEAN ENDOCRINOLOGY 2018; 14:57-61. [PMID: 30349595 PMCID: PMC6182922 DOI: 10.17925/ee.2018.14.2.57] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/03/2018] [Indexed: 12/13/2022]
Abstract
Acromegaly is a rare disease, caused largely by a growth hormone (GH) pituitary adenoma. Incidence is higher than previously thought. Due to increased morbidity and mortality, if not appropriately treated, early diagnosis efforts are essential. Screening is recommended for all patients with clinical features of GH excess. There is increased knowledge that classical diagnostic criteria no longer apply to all, and some patients can have GH excess with normal GH response to glucose. Treatment is multifactorial and personalised therapy is advised.
Collapse
Affiliation(s)
- Roula Zahr
- Department of Medicine, Division of Endocrinology, Diabetes and Clinical Nutrition
| | - Maria Fleseriu
- Department of Medicine, Division of Endocrinology, Diabetes and Clinical Nutrition
- Department of Neurological Surgery
- Northwest Pituitary Center, Oregon Health & Science University, Portland, OR, US
| |
Collapse
|
65
|
Melmed S, Bronstein MD, Chanson P, Klibanski A, Casanueva FF, Wass JAH, Strasburger CJ, Luger A, Clemmons DR, Giustina A. A Consensus Statement on acromegaly therapeutic outcomes. Nat Rev Endocrinol 2018; 14:552-561. [PMID: 30050156 PMCID: PMC7136157 DOI: 10.1038/s41574-018-0058-5] [Citation(s) in RCA: 371] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The 11th Acromegaly Consensus Conference in April 2017 was convened to update recommendations on therapeutic outcomes for patients with acromegaly. Consensus guidelines on the medical management of acromegaly were last published in 2014; since then, new pharmacological agents have been developed and new approaches to treatment sequencing have been considered. Thirty-seven experts in the management of patients with acromegaly reviewed the current literature and assessed changes in drug approvals, clinical practice standards and clinical opinion. They considered current treatment outcome goals with a focus on the impact of current and emerging somatostatin receptor ligands, growth hormone receptor antagonists and dopamine agonists on biochemical, clinical, tumour mass and surgical outcomes. The participants discussed factors that would determine pharmacological choices as well as the proposed place of each agent in the guidelines. We present consensus recommendations highlighting how acromegaly management could be optimized in clinical practice.
Collapse
Affiliation(s)
- Shlomo Melmed
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Marcello D Bronstein
- Division of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo, São Paulo, Brazil
| | - Philippe Chanson
- Assistance Publique-Hôpitaux de Paris, Service d'Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l'Hypophyse, Hôpital Bicêtre, Paris, France
- UMR S-1185, Faculté de Médecine Paris-Sud, Université Paris-Sud, Université Paris-Saclay, Paris, France
| | - Anne Klibanski
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Felipe F Casanueva
- Department of Medicine, Santiago de Compostela University, Santiago de Compostela, Spain
| | - John A H Wass
- Department of Endocrinology, Churchill Hospital, Oxford, UK
| | | | - Anton Luger
- Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - David R Clemmons
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Andrea Giustina
- Department of Endocrinology and Metabolism, San Raffaele University Hospital Milan, Milan, Italy
| |
Collapse
|
66
|
Mazziotti G, Frara S, Giustina A. Pituitary Diseases and Bone. Endocr Rev 2018; 39:440-488. [PMID: 29684108 DOI: 10.1210/er.2018-00005] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022]
Abstract
Neuroendocrinology of bone is a new area of research based on the evidence that pituitary hormones may directly modulate bone remodeling and metabolism. Skeletal fragility associated with high risk of fractures is a common complication of several pituitary diseases such as hypopituitarism, Cushing disease, acromegaly, and hyperprolactinemia. As in other forms of secondary osteoporosis, pituitary diseases generally affect bone quality more than bone quantity, and fractures may occur even in the presence of normal or low-normal bone mineral density as measured by dual-energy X-ray absorptiometry, making difficult the prediction of fractures in these clinical settings. Treatment of pituitary hormone excess and deficiency generally improves skeletal health, although some patients remain at high risk of fractures, and treatment with bone-active drugs may become mandatory. The aim of this review is to discuss the physiological, pathophysiological, and clinical insights of bone involvement in pituitary diseases.
Collapse
Affiliation(s)
| | - Stefano Frara
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Giustina
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
67
|
Frara S, Losa M, Doga M, Formenti AM, Mortini P, Mazziotti G, Giustina A. High Prevalence of Radiological Vertebral Fractures in Patients With TSH-Secreting Pituitary Adenoma. J Endocr Soc 2018; 2:1089-1099. [PMID: 30202830 PMCID: PMC6125713 DOI: 10.1210/js.2018-00091] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/24/2018] [Indexed: 01/10/2023] Open
Abstract
Background Bone loss and high risk of fractures have been reported in patients with primary hyperthyroidism, whereas data on skeletal health in TSH-secreting adenoma (TSH-oma) are scant, and the risk of fractures in this specific clinical context has not been investigated so far. In this cross-sectional study, we aimed at evaluating for the first time, to our knowledge, the prevalence and determinants of radiological vertebral fractures (VFs) in patients with TSH-oma. Methods Twenty-two patients (10 males, 12 females; median age 47 years) with TSH-oma and 44 patients (20 males, 24 females; median age 49 years) with nonfunctioning pituitary adenoma (NFPA) were retrospectively evaluated for thoracic VFs using a morphometric approach on lateral chest X-ray routinely performed in the presurgical diagnostic workup. Results The prevalence of VFs was significantly higher in TSH-oma vs NFPA (59.1% vs 22.7%; P = 0.003), the difference being still significant (odds ratio, 10.5; P = 0.005) after correction for the size of pituitary adenomas and biochemical parameters. In TSH-oma, the prevalence of VFs was significantly associated with older age (P = 0.007) and higher serum free T4 values (P = 0.02). In 20 patients, data on presurgical medical therapies of TSH-oma were available. All patients not treated with somatostatin receptor ligands were fractured compared with 25% of those who were treated with these drugs (P = 0.001). No significant (P = 0.25) association between VFs and treatment with methimazole was found. Conclusions This study provides the first evidence, to our knowledge, that patients with TSH-oma may develop VFs in close relationship with severity of hyperthyroidism.
Collapse
Affiliation(s)
- Stefano Frara
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| | - Marco Losa
- Institute of Neurosurgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Mauro Doga
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| | - Anna Maria Formenti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Pietro Mortini
- Institute of Neurosurgery, Università Vita-Salute San Raffaele, Milan, Italy
| | | | - Andrea Giustina
- Institute of Endocrinology, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
68
|
Mathews EH, Mathews GE, Meyer AA. A hypothetical method for controlling highly glycolytic cancers and metastases. Med Hypotheses 2018; 118:19-25. [PMID: 30037608 DOI: 10.1016/j.mehy.2018.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 01/23/2023]
Abstract
Most proliferating cancer cells and cancer-associated tumor stroma have an upregulated glucose energy demand in relation to normal cells. Cancer cells are further less metabolically flexible than normal cells. They can therefore not survive metabolic stress as well as normal cells can. Metabolic deprivation thus provides a potential therapeutic window. Unfortunately, current glucose blockers have toxicity problems. An alternative way to reduce a cancer patient's blood glucose (BG), for a short-term period to very low levels, without the concomitant toxicity, is hypothesized in this paper. In vitro tests have shown that short-term BG deprivation to 2 mmol/L for 180 min is an effective cancer treatment. This level of hypoglycaemia can be maintained in vivo with a combination of very low-dose insulin and the suppression of the glucose counter-regulation system. Such suppression can be safely achieved by the infusion of somatostatin and a combination of both α and β-blockers. The proposed short-term in vivo method, was shown to be non-toxic and safe for non-cancer patients. The next step is to test the effect of the proposed method on cancer patients. It is also suggested to incorporate well-known, long-term BG deprivation treatments to achieve maximum effect.
Collapse
Affiliation(s)
- Edward H Mathews
- CRCED, North-West University, P.O. Box 11207, Silver Lakes 0054, South Africa.
| | - George E Mathews
- CRCED, North-West University, P.O. Box 11207, Silver Lakes 0054, South Africa.
| | - Albertus A Meyer
- CRCED, North-West University, P.O. Box 11207, Silver Lakes 0054, South Africa.
| |
Collapse
|
69
|
Faggi L, Giustina A, Tulipano G. Effects of metformin on cell growth and AMPK activity in pituitary adenoma cell cultures, focusing on the interaction with adenylyl cyclase activating signals. Mol Cell Endocrinol 2018; 470:60-74. [PMID: 28962892 DOI: 10.1016/j.mce.2017.09.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 09/21/2017] [Accepted: 09/25/2017] [Indexed: 01/27/2023]
Abstract
For a few years we have been investigating AMP-activated protein kinase (AMPK) as a target for drug therapy of GH-secreting pituitary adenomas. Aim of this study was to investigate the direct effects of metformin, which causes AMPK activation in different cell types, on rat pituitary adenoma cell growth and on related cell signalling pathways. Our results suggest that metformin can exert a growth-inhibitory activity in rat pituitary tumor cells mediated by AMPK activation, although multiple mechanisms are most likely involved. Membrane proteins, including growth factor receptors, are valuable targets of AMPK. The inhibition of the mTOR-p70S6 kinase signalling pathway plays a role in the suppressive effect of metformin on pituitary tumor cell growth. Metformin did not affect the MTT reduction activity in energetic stress conditions. Finally, metformin was still able to induce AMPK activation and to inhibit cell growth in cells treated with forskolin and in transfected cells overexpressing GHRH-receptor and treated with GHRH. Hence, adenylyl cyclase over-activation does not account for the lack of response of some human pituitary tumors to AMPK-activating compounds in vitro.
Collapse
Affiliation(s)
- Lara Faggi
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Andrea Giustina
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Italy; Endocrine Service, University of Brescia, Italy
| | - Giovanni Tulipano
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Italy.
| |
Collapse
|
70
|
Leonart LP, Ferreira VL, Tonin FS, Fernandez-Llimos F, Pontarolo R. Medical Treatments for Acromegaly: A Systematic Review and Network Meta-Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2018; 21:874-880. [PMID: 30005760 DOI: 10.1016/j.jval.2017.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/28/2017] [Accepted: 12/11/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Acromegaly results from the hypersecretion of growth hormone. Because of the low incidence rates of this disease worldwide, few clinical trials evaluating drug treatments have been conducted. OBJECTIVES To conduct the first network meta-analysis simultaneously comparing all available drugs used in acromegaly treatment so as to provide more robust evidence in this field. METHODS A systematic review was performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses and Cochrane Collaboration recommendations (PROSPERO database under the registration number CRD42017059880). The electronic searches were conducted in PubMed (MEDLINE), Scopus, and Web of Science databases. Randomized controlled trials comparing any drug for the treatment of acromegaly head-to-head or versus placebo were included. Outcomes concerning the efficacy and safety of treatments were evaluated. The statistical analyses were performed using Aggregate Data Drug Information System version 1.16.8 (drugis.org, Groningen, The Netherlands). RESULTS The initial search retrieved 2059 articles. Of these, 10 randomized controlled trials were included in a qualitative analysis and 7 in a quantitative analysis. The network meta-analysis for the efficacy outcome (number of patients achieving insulinlike growth factor 1 control) showed that pegvisomant and lanreotide autogel were statistically superior to placebo (odds ratio [95% credible interval] 0.06 [0.00-0.55] and 0.09 [0.01-0.88]). No further differences were found. The probability rank indicated that pegvisomant and pasireotide have the highest probabilities (33% and 34%, respectively) of being the best therapeutic options. No major side effects were noted. CONCLUSIONS Pegvisomant is still a good option for acromegaly treatment, but pasireotide seems to be a promising alternative. Nevertheless, other important key factors such as drug costs and effectiveness (real-world results) should be taken into account when selecting acromegaly treatment.
Collapse
Affiliation(s)
- Letícia P Leonart
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Vinicius L Ferreira
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Fernanda S Tonin
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Fernando Fernandez-Llimos
- Department of Social Pharmacy, Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Roberto Pontarolo
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Paraná, Brazil.
| |
Collapse
|
71
|
Chiloiro S, Mazziotti G, Giampietro A, Bianchi A, Frara S, Mormando M, Pontecorvi A, Giustina A, De Marinis L. Effects of pegvisomant and somatostatin receptor ligands on incidence of vertebral fractures in patients with acromegaly. Pituitary 2018; 21:302-308. [PMID: 29397538 DOI: 10.1007/s11102-018-0873-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Acromegalic osteopathy is an emerging complication of acromegaly characterized by increase in bone turnover, deterioration in bone microarchitecture and high risk of vertebral fractures (VFs). Somatostatin receptor ligands (SRLs) and pegvisomant (PegV) are used for treatment of acromegaly and there is evidence that both drugs may exert direct effects on peripheral targets regardless of biochemical control of disease. However, whether or not SRLs and PegV may directly influence skeletal health its is unknown. METHODS In this longitudinal study, we evaluated the incidence of radiological VFs in 83 patients (48 females, 35 males; median age 47 years, range 18-80 years) who were treated with SRLs alone (42 cases), PegV alone (6 cases) or in combination with SRLs (35 cases) for median period of 82 months (range 36-126). PegV was given when acromegaly was not controlled by SRLs alone. RESULTS During the follow-up, 29 patients (34.9%) developed incident VFs. In patients receiving PegV due to active disease during SRL therapy, incidence of VFs decreased significantly from 43.9 to 26.8% (p = 0.039). When acromegaly was controlled by PegV, the incidence of VFs was slightly but not significantly lower as compared to that observed in patients with biochemical control of disease by SRLs (10.0 vs. 26.7%; p = 0.09). In the multivariate logistic regression analysis, incident VFs were independently predicted by pre-existing VFs (odds ratio 61.0; p = 0.009), duration of active acromegaly (odds ratio 1.01; p = 0.05) and mean serum IGF-I during the follow-up (odds ratio 5.26; p = 0.03), regardless of the therapeutic regimen (odds ratio 1.05; p = 0.94). CONCLUSIONS PegV and SRLs had comparable effects on VF risk in acromegaly. The activity of disease was the main determinant of VFs independently of the drug used to control acromegaly.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- Pituitary Unit, Catholic University of the Sacred Heart, Rome, Italy
| | | | | | - Antonio Bianchi
- Pituitary Unit, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Marilda Mormando
- Pituitary Unit, Catholic University of the Sacred Heart, Rome, Italy
| | | | | | - Laura De Marinis
- Pituitary Unit, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
72
|
Fedele M, Paciello O, De Biase D, Monaco M, Chiappetta G, Vitiello M, Barbieri A, Rea D, Luciano A, Papparella S, Arra C, Fusco A. HMGA2 cooperates with either p27 kip1 deficiency or Cdk4 R24C mutation in pituitary tumorigenesis. Cell Cycle 2018; 17:580-588. [PMID: 29157111 DOI: 10.1080/15384101.2017.1403682] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We have previously reported a critical role of HMGA proteins in pituitary tumorigenesis since either the Hmga1 or Hmga2 gene overexpression/activation induces the development of mixed growth hormone/prolactin cell pituitary adenomas by activating the E2F transcription factor 1, and then enhancing the G1/S transition of the cell cycle. Consistently, amplification and overexpression of the HMGA2 gene was found in human pituitary prolactinomas. Since impairment of the cell cycle control represents a feature of experimental and human pituitary adenomas, we have investigated the possible synergism between the alterations of other cell cycle regulators, such as p27 deficiency or Cdk4R24C mutation, with Hmga2 overexpression in pituitary tumorigenesis. Therefore, we crossed the Hmga2/T mice, overexpressing the truncated/active form of the Hmga2 gene, either with the knockout mice for p27kip1, or with the knockin mice for the Cdk4R24C mutation, both developing pituitary adenomas. Increased incidence and decreased latency in the development of pituitary lesions appeared in double mutant Hmga2/T;Cdk4R24C mice, and increased features of invasiveness and atypia were observed in pituitary tumors of both Hmga2/T;p27-ko and Hmga2/T;Cdk4R24C double mutant mice as compared with single mutant compounds. Interestingly, most of these mice develop pituitary adenomas with high Ki67 index, extrasellar expansion and brain tissue infiltration, representing good mouse models for human aggressive pituitary adenomas. Taken together, the results reported here indicate a cooperation between HMGA2 overexpression and either p27kip1 or CDK4 impairment in promoting pituitary tumor development and progression.
Collapse
Affiliation(s)
- Monica Fedele
- a CNR - Institute of Experimental Endocrinology and Oncology - c/o Department of Molecular Medicine and Medical Biotechnologies , University of Naples "Federico II" , Naples , Italy
| | - Orlando Paciello
- b Department of Veterinary Medicine and animal production , University of Naples "Federico II" , Naples , Italy
| | - Davide De Biase
- b Department of Veterinary Medicine and animal production , University of Naples "Federico II" , Naples , Italy
| | - Mario Monaco
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Gennaro Chiappetta
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Michela Vitiello
- a CNR - Institute of Experimental Endocrinology and Oncology - c/o Department of Molecular Medicine and Medical Biotechnologies , University of Naples "Federico II" , Naples , Italy
| | - Antonio Barbieri
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Domenica Rea
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Antonio Luciano
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Serenella Papparella
- b Department of Veterinary Medicine and animal production , University of Naples "Federico II" , Naples , Italy
| | - Claudio Arra
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Alfredo Fusco
- a CNR - Institute of Experimental Endocrinology and Oncology - c/o Department of Molecular Medicine and Medical Biotechnologies , University of Naples "Federico II" , Naples , Italy
| |
Collapse
|
73
|
Paragliola RM, Salvatori R. Novel Somatostatin Receptor Ligands Therapies for Acromegaly. Front Endocrinol (Lausanne) 2018; 9:78. [PMID: 29563895 PMCID: PMC5845985 DOI: 10.3389/fendo.2018.00078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Surgery is considered the treatment of choice in acromegaly, but patients with persistent disease after surgery or in whom surgery cannot be considered require medical therapy. Somatostatin receptor ligands (SRLs) octreotide (OCT), lanreotide, and the more recently approved pasireotide, characterized by a broader receptor ligand binding profile, are considered the mainstay in the medical management of acromegaly. However, in the attempt to offer a more efficacious and better tolerated medical approach, recent research has been aimed to override some limitations related to the use of currently approved drugs and novel SRLs therapies, with potential attractive features, have been proposed. These include both new formulation of older molecules and new molecules. Novel OCT formulations are aimed in particular to improve patients' compliance and to reduce injection discomfort. They include an investigational ready-to-use subcutaneous depot OCT formulation (CAM2029), delivered via prefilled syringes and oral OCT that uses a "transient permeability enhancer" technology, which allows for OCT oral absorption. Another new delivery system is a long-lasting OCT implant (VP-003), which provide stable doses of OCT throughout a period of several months. Finally, a new SRL DG3173 (somatoprim) seems to be more selective for GH secretion, suggesting possible advantages in the presence of hyperglycemia or diabetes. How much these innovations will actually be beneficial to acromegaly patients in real clinical practice remains to be seen.
Collapse
Affiliation(s)
| | - Roberto Salvatori
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, Pituitary Center Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Roberto Salvatori,
| |
Collapse
|
74
|
Nishioka H, Fukuhara N, Yamaguchi-Okada M, Takeshita A, Takeuchi Y, Yamada S. Pitfalls in early biochemical evaluation after transsphenoidal surgery in patients with acromegaly. Endocr J 2017; 64:1073-1078. [PMID: 28835593 DOI: 10.1507/endocrj.ej17-0261] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Although the current remission criteria for acromegaly are clear and concise, some pitfalls have been reported in early endocrinological evaluation after surgery. To evaluate the long-term (>4 year) outcome and to detect the pitfalls in early evaluation, we retrospectively reviewed 128 patients with acromegaly who underwent primary transsphenoidal surgery during 2011 and 2012. These included 66 men and 62 women, aged from 7 to 76 (mean 46) years old. 49 patients (38.3%) were preoperatively treated with somatostatin analog (SSA). Follow-up period ranged from 52 to 75 (63) months. Long-term remission using the current consensus criteria was achieved in 107 patients (83.6%), 105 of which patient had achieved remission in early evaluation. In 5 patients with preoperative SSA treatment, IGF-1 levels re-elevated more than one year after surgery. Five female patients without pretreatment with SSA showed delayed normalization of IGF-1 between 13 to 27 months postoperatively, two of which patients satisfied the remission criteria. In conclusion, the long-term results can be reliably predicted by the remission criteria early after surgery in most patients with acromegaly. For the accurate evaluation within a year after surgery, however, influence of preoperative treatment with SSA, delayed normalization of IGF-1, and poor GH suppression due to low insulin resistance must be considered, particularly in women.
Collapse
Affiliation(s)
- Hiroshi Nishioka
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, Tokyo, Japan
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Noriaki Fukuhara
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, Tokyo, Japan
| | | | - Akira Takeshita
- Department of Endocrinology, Toranomon Hospital, Tokyo, Japan
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Yasuhiro Takeuchi
- Department of Endocrinology, Toranomon Hospital, Tokyo, Japan
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Shozo Yamada
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, Tokyo, Japan
- Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| |
Collapse
|
75
|
A Case of Acromegaly in which a Pituitary Gland Tumor was Reduced Significantly by Administering Octreotide Long Acting Release (LAR) and Could Be Removed Surgically. J UOEH 2017; 39:241-245. [PMID: 28904276 DOI: 10.7888/juoeh.39.241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A 54-year-old woman was admitted to our hospital for detailed examination of acromegaly because she noticed bilateral hand and finger swelling at the age of 43 and plantar thickening, facial changes and unclear articulation at the age of 49. She had prominent brow ridges, mandibular protrusion, and enlargement of the hands, feet, nasal wings, lips and tongue. Her growth hormone (GH) level was 39.8 ng/ml, insulin-like growth factor-1 (IGF-1) level was 717 ng/ml, GH level was not suppressed (22.9 ng/ml) during a 75-g oral glucose tolerance test (OGTT). Radiography showed cauliflower-like enlargement of the distal phalanx of the fingers, thickening/enlargement of the plantar soft tissues, and increased antero-posterior diameter of the sella turcica. Magnetic resonance imaging showed a mass (21×17 mm) growing towards the right suprasellar region and invading the cavernous sinus. She was diagnosed with acromegaly based on the characteristic physical findings, GH excess, high IGF-1, lack of GH suppression during the 75-g OGTT, and the presence of a pituitary tumor. She was started on octreotide long acting release (Oct-LAR) 20 mg/4w for tumor shrinkage. After three doses, her GH and IGF-1 levels decreased to 2.19 ng/ml (1.69 during the 75-g OGTT) and 205 ng/ml, respectively, meeting cure criteria for acromegaly. In this case, a decrease in GH and IGF-1 levels, tumor shrinkage, and resolution of cavernous sinus invasion allowed the patient to undergo surgery with curative intent (the first-line treatment for acromegaly) without postoperative complications. Thus, preoperative Oct-LAR administration has the potential to improve treatment outcomes of acromegaly.
Collapse
|
76
|
Almeida JP, Ruiz-Treviño AS, Liang B, Omay SB, Shetty SR, Chen YN, Anand VK, Grover K, Christos P, Schwartz TH. Reoperation for growth hormone-secreting pituitary adenomas: report on an endonasal endoscopic series with a systematic review and meta-analysis of the literature. J Neurosurg 2017; 129:404-416. [PMID: 28862548 DOI: 10.3171/2017.2.jns162673] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Surgery is generally the first-line therapy for acromegaly. For patients with residual or recurrent tumors, several treatment options exist, including repeat surgery, medical therapy, and radiation. Reoperation for recurrent acromegaly has been associated with poor results, with hormonal control usually achieved in fewer than 50% of cases. Extended endonasal endoscopic approaches (EEAs) may potentially improve the results of reoperation for acromegaly by providing increased visibility and maneuverability in parasellar areas. METHODS A database of all patients treated in the authors' center between July 2004 and February 2016 was reviewed. Cases involving patients with acromegaly secondary to growth hormone (GH)-secreting adenomas who underwent EEA were selected for chart review and divided into 2 groups: first-time surgery and reoperation. Disease control was defined by 2010 guidelines. Clinical and radiological characteristics and outcome data were extracted. A systematic review was done through a MEDLINE database search (2000-2016) to identify studies on the surgical treatment of acromegaly. Using PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, the included studies were reviewed for surgical approach, tumor size, cavernous sinus invasion, disease control, and complications. Cases were divided into reoperation or first-time surgery for comparative analysis. RESULTS A total of 44 patients from the authors' institution were included in this study. Of these patients, 2 underwent both first-time surgery and reoperation during the study period and were therefore included in both groups. Thus data from 46 surgical cases were analyzed (35 first-time operations and 11 reoperations). The mean length of follow-up was 70 months (range 6-150 months). The mean size of the reoperated tumors was 14.8 ± 10.0 mm (5 micro- and 6 macroadenomas). The patients' mean age at the time of surgery was younger in the reoperation group than in the first-time surgery group (34.3 ± 12.8 years vs 49.1 ± 15.7 years, p = 0.007) and the mean preoperative GH level was also lower (7.7 ± 13.1 μg/L vs 25.6 ± 36.8 μg/L, p = 0.04). There was no statistically significant difference in disease control rates between the reoperation (7 [63.6%] of 11) and first-time surgery (25 [71.4%] of 33) groups (p = 0.71). Univariate analysis showed that older age, smaller tumor size, lower preoperative GH level, lower preoperative IGF-I level, and absence of cavernous sinus invasion were associated with higher chances of disease control in the first-time surgery group, whereas only absence of cavernous sinus invasion was associated with disease control in the reoperation group (p = 0.01). There was 1 case (9%) of transient diabetes insipidus and hypogonadism and 1 (9%) postoperative nasal infection after reoperation. The systematic review retrieved 29 papers with 161 reoperation and 2189 first-time surgery cases. Overall disease control for reoperation was 46.8% (95% CI 20%-74%) versus 56.4% (95% CI 49%-63%) for first-time operation. Reoperation and first-time surgery had similar control rates for microadenomas (73.6% [95% CI 32%-98%] vs 77.6% [95% CI 68%-85%]); however, reoperation was associated with substantially lower control rates for macroadenomas (27.5% [95% CI 5%-57%] vs 54.3% [95% CI 45%-62%]) and tumors invading the cavernous sinus (14.7% [95% CI 4%-29%] vs 38.5% [95% CI 27%-50%]). CONCLUSIONS Reoperative EEA for acromegaly had results similar to those for first-time surgery and rates of control for macroadenomas that were better than historical rates. Cavernous sinus invasion continues to be a negative prognostic indicator for disease control; however, results with EEA show improvement compared with results reported in the prior literature.
Collapse
|
77
|
Giustina A, Mazziotti G, Cannavò S, Castello R, Arnaldi G, Bugari G, Cozzi R, Ferone D, Formenti AM, Gatti E, Grottoli S, Maffei P, Maffezzoni F, Montini M, Terzolo M, Ghigo E. High-Dose and High-Frequency Lanreotide Autogel in Acromegaly: A Randomized, Multicenter Study. J Clin Endocrinol Metab 2017; 102:2454-2464. [PMID: 28419317 DOI: 10.1210/jc.2017-00142] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/12/2017] [Indexed: 02/13/2023]
Abstract
CONTEXT Increase in drug frequency or dose is recommended for acromegaly patients with partial response to long-acting somatostatin receptor ligands (SRLs). However, the efficacy and safety data with lanreotide (LAN) Autogel (LAN-ATG) at high dose (HD) or high frequency (HF) are still scanty. OBJECTIVE To evaluate the biochemical efficacy and safety of HF and HD LAN-ATG in patients with active acromegaly. DESIGN Twenty-four-week prospective, multicenter, randomized, open-label trial. PATIENTS AND INTERVENTIONS Thirty patients with active acromegaly, partial responders to SRLs, were randomized to HF (120 mg/21 days; 15 patients) or HD (180 mg/28 days; 15 patients) LAN-ATG. OUTCOMES Normalization of serum insulin-like growth factor-I (IGF-I) and reduction in random growth hormone (GH) values < 1.0 µg/L, reduction in serum IGF-I and GH from baseline, differences in biochemical response between HF and HD LAN-ATG, adverse events. RESULTS IGF-I decreased significantly (P = 0.007) during the 24-week treatment, with greater decrease in HD (P = 0.03) vs HF group (P = 0.08). Normalization in IGF-I values occurred in 27.6% of patients (P = 0.016 vs baseline), without a significant difference between HF and HD groups (P = 0.59). The decrease in serum IGF-I significantly correlated with serum LAN values (P = 0.04), and normalization of IGF-I was predicted by baseline IGF-I values (P = 0.02). Serum GH values did not change significantly (P = 0.22). Overall, 19 patients (63.3%) experienced adverse events, all being mild to moderate and transient, without differences between the two therapeutic arms. CONCLUSION HF and HD LAN-ATG regimens are effective in normalizing IGF-I values in about one-third of patients with active acromegaly inadequately controlled by long-term conventional SRLs therapy.
Collapse
Affiliation(s)
- Andrea Giustina
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Gherardo Mazziotti
- Endocrinology Unit, Azienda Socio Sanitaria Territoriale Carlo Poma, 46100 Mantova, Italy
| | | | - Roberto Castello
- Division of General Medicine, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy
| | - Giorgio Arnaldi
- Division of Endocrinology, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Giovanna Bugari
- Laboratory of Chemical Analysis, Azienda Socio Sanitaria Territoriale Spedali Civili, 25126 Brescia, Italy
| | - Renato Cozzi
- Division of Endocrinology, Ospedale Niguarda, 20162 Milan, Italy
| | - Diego Ferone
- Endocrinology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto dei Tumori, University of Genoa, 16132 Genoa, Italy
| | - Anna Maria Formenti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Enza Gatti
- Neuroradiology, University of Brescia, 25123 Brescia, Italy
| | - Silvia Grottoli
- Division of Endocrinology and Metabolism, University of Turin, 10126 Turin, Italy
| | - Pietro Maffei
- Department of Medicine, Padua University Hospital, 35121 Padua, Italy
| | - Filippo Maffezzoni
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | | | | | - Ezio Ghigo
- Division of Endocrinology and Metabolism, University of Turin, 10126 Turin, Italy
| |
Collapse
|
78
|
Surgical treatment and outcome of TSH-producing pituitary adenomas. Acta Neurochir (Wien) 2017; 159:1219-1226. [PMID: 28204898 DOI: 10.1007/s00701-017-3105-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Thyrotropin-producing adenomas (TSHomas) account for approximately 1-2% of all pituitary tumors. Recently, there has been debate on primary treatment as some studies suggest a high rate of hypopituitarism after transsphenoidal surgery and therefore suggest primary use of somatostatin analogs (SSA) instead. We would like to challenge this assumption by presenting our single-center experience with transsphenoidal surgery for TSHoma. METHODS Fifteen patients treated consecutively between May 2010 and December 2016 were analyzed for long-term postoperative remission and pituitary function. RESULTS Data on the development of TSH, fT3 and fT4 were available for 12 of 15 patients (mean follow-up was 18 months, 8 patients >12 months), showing mean TSH levels of 1.289 mU/l (0.02-2.04, SD 0.66), mean fT3 levels of 3.76 pmol/l (0.5-6.16, SD 1.8) and mean fT4 levels of 16.5 pmol/l (11.7-21.9, SD 3.66). Six of those patients were substituted with a mean of 85.4 μg L-thyroxine after a median follow-up of 20.7 months. The other six patients did not receive L-thyroxine at a median follow-up of 15.5 months. One patient with a known tumor remnant on MRI stayed euthyreotic with cabergoline at the timepoint of follow-up 22 months after the operation. Control of the corticotrop axis was also available in 12 of 15 with no patient showing a corticotroph insufficiency in the long term. CONCLUSIONS We argue that transsphenoidal surgery for TSHoma should be considered as the treatment of choice as remission following surgery is highly probable and postoperative hypopituitarism is very unlikely if patients are referred to centers with high pituitary surgery case loads.
Collapse
|
79
|
Langlois F, McCartney S, Fleseriu M. Recent Progress in the Medical Therapy of Pituitary Tumors. Endocrinol Metab (Seoul) 2017; 32:162-170. [PMID: 28685507 PMCID: PMC5503860 DOI: 10.3803/enm.2017.32.2.162] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
Management of pituitary tumors is multidisciplinary, with medical therapy playing an increasingly important role. With the exception of prolactin-secreting tumors, surgery is still considered the first-line treatment for the majority of pituitary adenomas. However, medical/pharmacological therapy plays an important role in controlling hormone-producing pituitary adenomas, especially for patients with acromegaly and Cushing disease (CD). In the case of non-functioning pituitary adenomas (NFAs), pharmacological therapy plays a minor role, the main objective of which is to reduce tumor growth, but this role requires further studies. For pituitary carcinomas and atypical adenomas, medical therapy, including chemotherapy, acts as an adjuvant to surgery and radiation therapy, which is often required to control these aggressive tumors. In the last decade, knowledge about the pathophysiological mechanisms of various pituitary adenomas has increased, thus novel medical therapies that target specific pathways implicated in tumor synthesis and hormonal over secretion are now available. Advancement in patient selection and determination of prognostic factors has also helped to individualize therapy for patients with pituitary tumors. Improvements in biochemical and "tumor mass" disease control can positively affect patient quality of life, comorbidities and overall survival. In this review, the medical armamentarium for treating CD, acromegaly, prolactinomas, NFA, and carcinomas/aggressive atypical adenomas will be presented. Pharmacological therapies, including doses, mode of administration, efficacy, adverse effects, and use in special circumstances are provided. Medical therapies currently under clinical investigation are also briefly discussed.
Collapse
Affiliation(s)
- Fabienne Langlois
- Department of Medicine, Endocrinology and Metabolism, University of Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
- Northwest Pituitary Center, Oregon Health & Science University, Portland, OR, USA
| | - Shirley McCartney
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
- Northwest Pituitary Center, Oregon Health & Science University, Portland, OR, USA
| | - Maria Fleseriu
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
- Northwest Pituitary Center, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
80
|
Giustina A, Arnaldi G, Bogazzi F, Cannavò S, Colao A, De Marinis L, De Menis E, Degli Uberti E, Giorgino F, Grottoli S, Lania AG, Maffei P, Pivonello R, Ghigo E. Pegvisomant in acromegaly: an update. J Endocrinol Invest 2017; 40:577-589. [PMID: 28176221 PMCID: PMC5443862 DOI: 10.1007/s40618-017-0614-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND In 2007, we published an opinion document to review the role of pegvisomant (PEG) in the treatment of acromegaly. Since then, new evidence emerged on the biochemical and clinical effects of PEG and on its long-term efficacy and safety. AIM We here reviewed the emerging aspects of the use of PEG in clinical practice in the light of the most recent literature. RESULTS The clinical use of PEG is still suboptimal, considering that it remains the most powerful tool to control IGF-I in acromegaly allowing to obtain, with a pharmacological treatment, the most important clinical effects in terms of signs and symptoms, quality of life and comorbidities. The number of patients with acromegaly exposed to PEG worldwide has become quite elevated and the prolonged follow-up allows now to deal quite satisfactorily with many clinical issues including major safety issues, such as the concerns about possible tumour (re)growth under PEG. The positive or neutral impact of PEG on glucose metabolism has been highlighted, and the clinical experience, although limited, with sleep apnoea and pregnancy has been reviewed. Finally, the current concept of somatostatin receptor ligands (SRL) resistance has been addressed, in order to better define the acromegaly patients to whom the PEG option may be offered. CONCLUSIONS PEG increasingly appears to be an effective and safe medical option for many patients not controlled by SRL but its use still needs to be optimized.
Collapse
Affiliation(s)
- A Giustina
- Chair of Endocrinology, Vita-Salute San Raffaele University, Milano, Italy.
| | - G Arnaldi
- Clinic of Endocrinology and Metabolism Disease, Ospedali Riuniti di Ancona, Ancona, Italy
| | - F Bogazzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Cannavò
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - A Colao
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - L De Marinis
- Pituitary Unit, Catholic University School of Medicine, Rome, Italy
| | - E De Menis
- Department of Internal Medicine, General Hospital, Montebelluna (TV), Italy
| | - E Degli Uberti
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - F Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - S Grottoli
- Endocrinology, Diabetology and Metabolism, AOU Città della Salute e della Scienza of Turin, Turin, Italy
| | - A G Lania
- Endocrinology Unit, Department of Biomedical Sciences, Humanitas University and Humanitas Research Hospital, Rozzano, Italy
| | - P Maffei
- Department of Medicine (DIMED), 3rd Medical Clinic, Azienda Ospedaliera Padova, Padova, Italy
| | - R Pivonello
- Department of Clinical and Surgery Medicine, Endocrinology and Metabolism, University of Naples, Naples, Italy
| | - E Ghigo
- Department of Medical Sciences, School of Medicine, University of Turin, Turin, Italy
| |
Collapse
|
81
|
Eroukhmanoff J, Tejedor I, Potorac I, Cuny T, Bonneville JF, Dufour H, Weryha G, Beckers A, Touraine P, Brue T, Castinetti F. MRI follow-up is unnecessary in patients with macroprolactinomas and long-term normal prolactin levels on dopamine agonist treatment. Eur J Endocrinol 2017; 176:323-328. [PMID: 28073906 DOI: 10.1530/eje-16-0897] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/21/2016] [Accepted: 01/10/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Both antitumor and antisecretory efficacies of dopamine agonists (DA) make them the first-line treatment of macroprolactinomas. However, there is no guideline for MRI follow-up once prolactin is controlled. The aim of our study was to determine whether a regular MRI follow-up was necessary in patients with long-term normal prolactin levels under DA. PATIENTS AND METHODS We conducted a retrospective multicenter study (Marseille, Paris La Pitie Salpetriere and Nancy, France; Liege, Belgium) including patients with macroprolactinomas (largest diameter: >10 mm and baseline prolactin level: >100 ng/mL) treated by dopamine agonists, and regularly followed (pituitary MRI and prolactin levels) during at least 48 months once normal prolactin level was obtained. RESULTS In total, 115 patients were included (63 men and 52 women; mean age at diagnosis: 36.3 years). Mean baseline prolactin level was 2224 ± 6839 ng/mL. No significant increase of tumor volume was observed during the follow-up. Of the 21 patients (18%) who presented asymptomatic hemorrhagic changes of the macroprolactinoma on MRI, 2 had a tumor increase (2 and 7 mm in the largest size). Both were treated by cabergoline (1 mg/week) with normal prolactin levels obtained for 6 and 24 months. For both patients, no further growth was observed on MRI during follow-up at the same dose of cabergoline. CONCLUSION No significant increase of tumor size was observed in our patients with controlled prolactin levels on DA. MRI follow-up thus appears unnecessary in patients with biologically controlled macroprolactinomas.
Collapse
Affiliation(s)
- J Eroukhmanoff
- Aix Marseille UniversityAssistance Publique Hopitaux de Marseille, La Conception Hospital, Marseille, France
| | - I Tejedor
- Groupe Hospitalier Pitié-Salpêtrière Service d'Endocrinologie & Médecine de la ReproductionParis, France
| | - I Potorac
- Domaine Universitaire du Sart Tilman CHU de Liège Service d'EndocrinologieLiege, Belgium
| | - T Cuny
- CHU de Nancy - Hôpital de Brabois Clinique Médicale et EndocrinologiqueNancy, France
| | - J F Bonneville
- Domaine Universitaire du Sart Tilman CHU de Liège Service d'EndocrinologieLiege, Belgium
| | - H Dufour
- Service de NeurochirurgieHôpital de la Timone, Marseille, France
| | - G Weryha
- CHU de Nancy - Hôpital de Brabois Clinique Médicale et EndocrinologiqueNancy, France
| | - A Beckers
- Domaine Universitaire du Sart Tilman CHU de Liège Service d'EndocrinologieLiege, Belgium
| | - P Touraine
- Groupe Hospitalier Pitié-Salpêtrière Service d'Endocrinologie & Médecine de la ReproductionParis, France
| | - T Brue
- Aix Marseille UniversityAssistance Publique Hopitaux de Marseille, La Conception Hospital, Marseille, France
| | - F Castinetti
- Aix Marseille UniversityAssistance Publique Hopitaux de Marseille, La Conception Hospital, Marseille, France
| |
Collapse
|
82
|
Abstract
Aggressive GH-secreting pituitary adenomas (GHPAs) represent an important clinical problem in patients with acromegaly. Surgical therapy, although often the mainstay of treatment for GHPAs, is less effective in aggressive GHPAs due to their invasive and destructive growth patterns, and their proclivity for infrasellar invasion. Medical therapies for GHPAs, including somatostatin analogues and GH receptor antagonists, are becoming increasingly important adjuncts to surgical intervention. Stereotactic radiosurgery serves as an important fallback therapy for tumors that cannot be cured with surgery and medications. Data suggests that patients with aggressive and refractory GHPAs are best treated at dedicated tertiary pituitary centers with multidisciplinary teams of neuroendocrinologists, neurosurgeons, radiation oncologists and other specialists who routinely provide advanced care to GHPA patients. Future research will help clarify the defining features of "aggressive" and "atypical" PAs, likely based on tumor behavior, preoperative imaging characteristics, histopathological characteristics, and molecular markers.
Collapse
Affiliation(s)
- Daniel A Donoho
- Department of Neurological Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Namrata Bose
- Division of Endocrinology, Department of Medicine, Keck School of Medicine of the University of Southern California, USC Pituitary Center, 1520 San Pablo Street #3800, Los Angeles, CA, 90033, USA
| | - Gabriel Zada
- Department of Neurological Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - John D Carmichael
- Division of Endocrinology, Department of Medicine, Keck School of Medicine of the University of Southern California, USC Pituitary Center, 1520 San Pablo Street #3800, Los Angeles, CA, 90033, USA.
| |
Collapse
|
83
|
Paragliola RM, Corsello SM, Salvatori R. Somatostatin receptor ligands in acromegaly: clinical response and factors predicting resistance. Pituitary 2017; 20:109-115. [PMID: 27778296 DOI: 10.1007/s11102-016-0768-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Somatostatin (SST) receptor ligands (SRL), in particular those of first generation (Octreotide and Lanreotide), are widely used in medical treatment of acromegaly, but they assure biochemical control of disease (and the possibility of an improvement of clinical symptoms and tumor shrinkage), only in a subset of patients. DISCUSSION The mechanisms underlying the so called "SRL resistance" are various and involve in particular SST receptor expression and molecular pathways of signal transduction. Different predictors of SRL response have been reported, including clinical and biochemical features (gender, age, growth hormone and insulin-like growth factor-I levels at diagnosis), and tumor characteristic (both at preoperative magnetic resonance imaging study and histopathology) as well as expression of SST receptors. In some cases, only a "partial resistance" to SST can be detected, probably due to the presence of other impaired molecular mechanisms involved in signal transduction, which compromise specific pathways and not others. This may explain some cases of dissociated response between biochemical control and tumor shrinkage.
Collapse
Affiliation(s)
- Rosa Maria Paragliola
- Unit of Endocrinology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Salvatore Maria Corsello
- Unit of Endocrinology, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Roberto Salvatori
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Pituitary Center, Johns Hopkins University School of Medicine, 1830 East Monument Street Suite #333, Baltimore, MD, 21287, USA.
| |
Collapse
|
84
|
Abstract
Acromegalic osteopathy is an emerging complication of acromegaly characterized by increase in bone turnover, deterioration in bone microarchitecture and high risk of vertebral fractures. Vertebral fractures, as diagnosed by a radiological and morphometric approach, occur in about one-third of acromegaly patients in close relationship with duration of active disease. However, the prediction of vertebral fractures in this clinical setting is still a matter of uncertainty, since the pathogenesis of acromegalic osteopathy is multifactorial and fractures may occur even in presence of normal bone mineral density. In this narrative article, we summarize the pathophysiology and clinical aspects of acromegalic osteopathy.
Collapse
Affiliation(s)
- G Mazziotti
- Chair of Endocrinology, University of Brescia, Via Biseo, 17, 25123, Brescia, Italy
- Endocrinology Unit, ASST Carlo Poma of Mantua, Mantua, Italy
| | - F Maffezzoni
- Chair of Endocrinology, University of Brescia, Via Biseo, 17, 25123, Brescia, Italy
| | - S Frara
- Chair of Endocrinology, University of Brescia, Via Biseo, 17, 25123, Brescia, Italy
| | - A Giustina
- Chair of Endocrinology, University of Brescia, Via Biseo, 17, 25123, Brescia, Italy.
| |
Collapse
|
85
|
Formenti AM, Maffezzoni F, Doga M, Mazziotti G, Giustina A. Growth hormone deficiency in treated acromegaly and active Cushing's syndrome. Best Pract Res Clin Endocrinol Metab 2017; 31:79-90. [PMID: 28477735 DOI: 10.1016/j.beem.2017.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Growth hormone deficiency (GHD) in adults is characterized by reduced quality of life and physical fitness, skeletal fragility, increased weight and cardiovascular risk. It may be found in (over-) treated acromegaly as well as in active Cushing's syndrome. Hypopituitarism may develop in patients after definitive treatment of acromegaly, although the exact prevalence of GHD in this population is still uncertain because of limited awareness, and scarce and conflicting data so far available. Because GHD associated with acromegaly and Cushing's syndrome may yield adverse consequences on similar target systems, the final outcomes of some complications of both acromegaly and Cushing's syndrome may be further affected by the occurrence of GHD. It is still largely unknown, however, whether GHD in patients with post-acromegaly or active Cushing's syndrome (e.g. pharmacologic glucocorticoid treatment) may benefit from GH replacement. We review the diagnostic, clinical and therapeutic aspects of GHD in adults treated for acromegaly and in those with active Cushing's syndrome.
Collapse
Affiliation(s)
| | | | - Mauro Doga
- Endocrinology, Univeristy of Brescia, Italy
| | | | | |
Collapse
|
86
|
Abstract
PURPOSE Uncontrolled acromegaly results in approximately 2-fold excess mortality. Pituitary surgery is first-line therapy, and medical treatment is indicated for persistent disease. While cabergoline and pegvisomant are used in select patients, somatostatin receptor ligands (SRLs) remain the cornerstone of medical treatment. Management of patients poorly responsive to SRLs is therefore, challenging. The purpose of this review is to highlight the options for combination medical therapy in the treatment of acromegaly, with an emphasis on efficacy and safety. METHODS All original articles/abstracts detailing combination medical therapy in acromegaly were identified from a PubMed search. RESULTS Studies reviewed included retrospective and open-label prospective studies. While the combination of SRL and cabergoline was generally well tolerated, a lower baseline insulin-like growth factor-1 (IGF-1) level was the best predictor of efficacy; this combination may be most effective in patients with mildly elevated IGF-1. SRL-pegvisomant combination normalized IGF-1 in the majority of patients; continued efficacy despite individual drug dosing reduction was also reported. The risk of significant liver enzyme elevation was, however, higher than that reported with SRL monotherapy; close monitoring is recommended. Data on pegvisomant-cabergoline combination is limited, but this may be an option in the setting of SRL intolerance. Reports on temozolomide used in combination with other medical therapies in patients with aggressive GH-secreting tumors are also summarized. CONCLUSION While more prospective, randomized controlled trials on long-term efficacy and safety are needed, combination medical therapy remains a treatment strategy that should be considered for acromegaly patients poorly responsive to SRLs.
Collapse
Affiliation(s)
- Dawn Shao Ting Lim
- Departments of Medicine (Endocrinology) and Neurological Surgery, and Northwest Pituitary Center, Oregon Health & Science University, 3303 SW Bond Ave, Mail Code CH8N, Portland, OR, 97239, USA
| | - Maria Fleseriu
- Departments of Medicine (Endocrinology) and Neurological Surgery, and Northwest Pituitary Center, Oregon Health & Science University, 3303 SW Bond Ave, Mail Code CH8N, Portland, OR, 97239, USA.
| |
Collapse
|
87
|
Hong GK. Medical Management for Pituitary Adenoma Patients. TRANSSPHENOIDAL SURGERY 2017:359-382. [DOI: 10.1007/978-3-319-56691-7_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
88
|
Grasso LFS, Auriemma RS, Pivonello R, Colao A. Somatostatin analogs, cabergoline and pegvisomant: comparing the efficacy of medical treatment for acromegaly. Expert Rev Endocrinol Metab 2017; 12:73-85. [PMID: 30058878 DOI: 10.1080/17446651.2016.1222899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Therapies for acromegaly aim at normalizing hormonal excess and controlling tumor growth . Therapeutic approaches are surgery, pharmacotherapy and radiotherapy. Area covered: This review focuses on the role of medical therapy of acromegaly, comparing the efficacy of somatostatin analogues (SSA), dopamine-agonists (DA) and pegvisomant (PEG), the three available drug classes for treating acromegaly. To clarify the difference in response rates reported in the literature for these therapies, we performed a search for original articles published in PubMed. SSA represent the first-line approach to medical treatment. This therapy is effective in controlling acromegaly in about 40% of patients, however there are great differences in the reported hormonal efficacy of SSA in the different series. In patients partially resistant to SSA, cabergoline can be added when hormonal levels are close to normalization, resulting effective in control IGF-I levels in 43% of patients. In patients with higher hormonal levels PEG is indicated, normalizing IGF-I levels in 79.8% and 80.6% of cases when used in monotherapy or in combination with SSA. Pasireotide, the newly developed SSA multi-ligand receptor, represents a new option in SSA resistant patients. Expert commentary: Medical therapy represents an important therapeutic option resulting safe and effective in controlling acromegaly in a high percentage of patients. The best treatment should be individually tailored for each patient, taking into account sex, age, comorbidities, tumor characteristics and hormonal levels.
Collapse
Affiliation(s)
- Ludovica F S Grasso
- a Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia , 'Federico II' University of Naples , Naples , Italy
| | - Renata S Auriemma
- a Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia , 'Federico II' University of Naples , Naples , Italy
| | - Rosario Pivonello
- a Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia , 'Federico II' University of Naples , Naples , Italy
| | - Annamaria Colao
- a Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia , 'Federico II' University of Naples , Naples , Italy
| |
Collapse
|
89
|
Kirzinger L, Boy S, Marienhagen J, Schuierer G, Neu R, Ried M, Hofmann HS, Wiebe K, Ströbel P, May C, Kleylein-Sohn J, Baierlein C, Bogdahn U, Marx A, Schalke B. Octreotide LAR and Prednisone as Neoadjuvant Treatment in Patients with Primary or Locally Recurrent Unresectable Thymic Tumors: A Phase II Study. PLoS One 2016; 11:e0168215. [PMID: 27992479 PMCID: PMC5161359 DOI: 10.1371/journal.pone.0168215] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/23/2016] [Indexed: 12/17/2022] Open
Abstract
Therapeutic options to cure advanced, recurrent, and unresectable thymomas are limited. The most important factor for long-term survival of thymoma patients is complete resection (R0) of the tumor. We therefore evaluated the response to and the induction of resectability of primarily or locally recurrent unresectable thymomas and thymic carcinomas by octreotide Long-Acting Release (LAR) plus prednisone therapy in patients with positive octreotide scans. In this open label, single-arm phase II study, 17 patients with thymomas considered unresectable or locally recurrent thymoma (n = 15) and thymic carcinoma (n = 2) at Masaoka stage III were enrolled. Octreotide LAR (30 mg once every 2 weeks) was administered in combination with prednisone (0.6 mg/kg per day) for a maximum of 24 weeks (study design according to Fleming´s one sample multiple testing procedure for phase II clinical trials). Tumor size was evaluated by volumetric CT measurements, and a decrease in tumor volume of at least 20% at week 12 compared to baseline was considered as a response. We found that octreotide LAR plus prednisone elicited response in 15 of 17 patients (88%). Median reduction of tumor volume after 12 weeks of treatment was 51% (range 20%-86%). Subsequently, complete surgical resection was achieved in five (29%) and four patients (23%) after 12 and 24 weeks, respectively. Octreotide LAR plus prednisone treatment was discontinued in two patients before week 12 due to unsatisfactory therapeutic effects or adverse events. The most frequent adverse events were gastrointestinal (71%), infectious (65%), and hematological (41%) complications. In conclusion, octreotide LAR plus prednisone is efficacious in patients with primary or recurrent unresectable thymoma with respect to tumor regression. Octreotide LAR plus prednisone was well tolerated and adverse events were in line with the known safety profile of both agents.
Collapse
Affiliation(s)
- Lukas Kirzinger
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Sandra Boy
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Jörg Marienhagen
- Department of Nuclear Medicine, University of Regensburg, Regensburg, Germany
| | | | - Reiner Neu
- Department of Thoracic Surgery, University of Regensburg, Regensburg, Germany
| | - Michael Ried
- Department of Thoracic Surgery, University of Regensburg, Regensburg, Germany
| | - Hans-Stefan Hofmann
- Department of Thoracic Surgery, University of Regensburg, Regensburg, Germany
| | - Karsten Wiebe
- Department of Cardiac and Thoracic Surgery, University of Muenster, Muenster, Germany
| | - Philipp Ströbel
- Institute of Pathology, University of Goettingen, Goettingen, Germany
| | | | | | | | - Ulrich Bogdahn
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Berthold Schalke
- Department of Neurology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
90
|
Öberg K, Lamberts SWJ. Somatostatin analogues in acromegaly and gastroenteropancreatic neuroendocrine tumours: past, present and future. Endocr Relat Cancer 2016; 23:R551-R566. [PMID: 27697899 DOI: 10.1530/erc-16-0151] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/03/2016] [Indexed: 11/08/2022]
Abstract
Acromegaly is a hormonal disorder that arises when the pituitary gland secretes excess growth hormone (GH), which in turn stimulates a concomitant increase in serum insulin-like growth factor 1 (IGF-1) levels. Gastroenteropancreatic neuroendocrine tumours (GEP-NET) constitute a heterogeneous group of tumours that can secrete serotonin and a variety of peptide hormones that may cause characteristic symptoms known as carcinoid syndrome or other symptoms and hormonal hypersecretion syndromes depending on the tumour's site of origin. Current medical therapy for the treatment of acromegaly and GEP-NET involves the administration of somatostatin analogues that effectively suppress excess hormone secretion. After its discovery in 1979, octreotide became the first synthetic biologically stable somatostatin analogue with a short-acting formulation of octreotide introduced into clinical practice in the late 1980s. Lanreotide, another somatostatin analogue, became available in the mid-1990s initially as a prolonged-release formulation administered every 10 or 14 days. Long-acting release formulations of both octreotide (Sandostatin LAR and Novartis) and lanreotide (Somatuline Autogel, Ipsen), based on microparticle and nanoparticle drug-delivery technologies, respectively, were later developed, which allowed for once-monthly administration and improved convenience. First-generation somatostatin analogues remain one of the cornerstones of medical therapy in the management of pituitary and GEP-NET hormone hypersecretion, with octreotide having the longest established efficacy and safety profile of the somatostatin analogue class. More recently, pasireotide (Signifor), a next-generation multireceptor-targeted somatostatin analogue, has emerged as an alternative therapeutic option for the treatment of acromegaly. This review summarizes the development and clinical success of somatostatin analogues.
Collapse
|
91
|
Maffezzoni F, Frara S, Doga M, Mazziotti G, Giustina A. New medical therapies of acromegaly. Growth Horm IGF Res 2016; 30-31:58-63. [PMID: 27745780 DOI: 10.1016/j.ghir.2016.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 09/26/2016] [Accepted: 10/02/2016] [Indexed: 12/29/2022]
Abstract
Acromegaly is a rare disease associated with significant morbidity and increased mortality. Treatment of acromegaly aims at controlling growth hormone hypersecretion, improving patients' symptoms and comorbidities and normalizing mortality. The therapeutic options for acromegaly include surgery, medical therapies and radiotherapy. However, despite all these treatment options, approximately one-half of patients are not adequately controlled. Progress in molecular research has made possible to develop new therapeutic strategies to improve control of acromegaly. This article will review the new medical approaches to acromegaly which consist in evolution of traditional therapeutic protocols and development of new molecules with different profiles of activity.
Collapse
Affiliation(s)
| | - S Frara
- Endocrinology, University of Brescia, Italy
| | - M Doga
- Endocrinology, University of Brescia, Italy
| | - G Mazziotti
- Endocrinology, University of Brescia, Italy; Endocrinology, ASST Carlo Poma of Mantua, Italy
| | - A Giustina
- Endocrinology, University of Brescia, Italy.
| |
Collapse
|
92
|
Waligórska-Stachura J, Gut P, Sawicka-Gutaj N, Liebert W, Gryczyńska M, Baszko-Błaszyk D, Blanco-Gangoo AR, Ruchała M. Growth hormone–secreting macroadenoma of the pituitary gland successfully treated with the radiolabeled somatostatin analog 90Y-DOTATATE: case report. J Neurosurg 2016; 125:346-9. [DOI: 10.3171/2015.6.jns15363] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pituitary tumors causing acromegaly are usually macroadenomas at the time of diagnosis, and they can grow aggressively, infiltrating surrounding tissues. Difficulty in achieving complete tumor removal at surgery can lead toward a strong tendency for recurrence, making it necessary to consider a means of treatment other than those currently used such as somatostatin analogs (SSAs), growth hormone (GH) receptor antagonist, surgical removal, and radiotherapy. The purpose of this paper is to describe a patient diagnosed with an aggressive, giant GH-secreting tumor refractory to medical therapy but ultimately treated with the radiolabeled somatostatin analog 90Y-DOTATATE.
A 26-year-old male with an invasive macroadenoma of the pituitary gland (5.6 × 2.5 × 3.6 cm) and biochemically confirmed acromegaly underwent 2 partial tumor resections: the first used the transsphenoidal approach and the second used the transcranial method. The patient received SSAs pre- and postoperatively. Because of the progression in pituitary tumor size, he underwent classic irradiation of the tumor (50 Gy). One and a half years later, the patient presented with clinically and biochemically active disease, and the tumor size was still 52 mm in diameter (height). Two neurosurgeons disqualified him from further surgical procedures. After confirming the presence of somatostatin receptors in the pituitary tumor by using 68Ga-DOTATATE PET/CT, we treated the patient 4 times with an SSA bound with 90Y-DOTATATE. After this treatment, the patient attained partial biochemical remission and a reduction in the tumor mass for the first time.
Treatment with an SSA bound with 90Y-DOTATATE may be a promising option for some aggressive GH-secreting pituitary adenomas when other methods have failed.
Collapse
Affiliation(s)
| | - Paweł Gut
- Departments of 1Endocrinology, Metabolism and Internal Medicine and
| | | | - Włodzimierz Liebert
- 2Neurosurgery and Neurotraumatology, Poznań University of Medical Sciences, Poznań, Poland
| | - Maria Gryczyńska
- Departments of 1Endocrinology, Metabolism and Internal Medicine and
| | | | | | - Marek Ruchała
- Departments of 1Endocrinology, Metabolism and Internal Medicine and
| |
Collapse
|
93
|
Schilbach K, Schopohl J. Update on the use of oral octreotide therapy for acromegaly. Expert Rev Endocrinol Metab 2016; 11:349-355. [PMID: 30058923 DOI: 10.1080/17446651.2016.1199954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Somatostatin analogs are most commonly used in pharmacological treatment of acromegaly. Pegvisomant and dopamine agonists are alternatives, which are used to a lesser extent. Dopamine agonists are the only orally applicable medication but are less effective than the other options. For a large number of patients, life-long pharmacotherapy has to be applied and frequent injections represent a reduction of quality of life for many of them. Areas covered: Recently published evidence for the use of oral octreotide therapy for acromegaly. Expert commentary: Oral octreotide is a novel and effective treatment for acromegaly and the side effects have been shown to be comparable to the injectable SSAs. The combination with a transient permeability enhancer allows intestinal permeation but also enables molecules with a size <70 kDa to pass transiently. This does not seem to have an acute or subacute consequence, but the long-term effect is still elusive. Therefore, more long-term trials are desirable.
Collapse
Affiliation(s)
- Katharina Schilbach
- a Medizinische Klinik und Poliklinik IV , Klinikum der Universität München , Munich , Germany
| | - Jochen Schopohl
- a Medizinische Klinik und Poliklinik IV , Klinikum der Universität München , Munich , Germany
| |
Collapse
|
94
|
Pisani A, Sabbatini M, Imbriaco M, Riccio E, Rubis N, Prinster A, Perna A, Liuzzi R, Spinelli L, Santangelo M, Remuzzi G, Ruggenenti P. Long-term Effects of Octreotide on Liver Volume in Patients With Polycystic Kidney and Liver Disease. Clin Gastroenterol Hepatol 2016; 14:1022-1030.e4. [PMID: 26844873 DOI: 10.1016/j.cgh.2015.12.049] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 12/17/2015] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS Short-term studies have shown that somatostatin analogues are effective in patients with polycystic kidney and liver disease. We evaluated the long-term effects of long-acting release octreotide (octreotide LAR), a somatostatin inhibitor, vs placebo in these patients. METHODS We performed a controlled study of adults with polycystic kidney and liver disease (estimated glomerular filtration rate, 40 mL/min/1.73m(2) or more) at a single center in Italy. We analyzed data from 27 patients randomly assigned to groups given octreotide LAR (40 mg, n = 14) or placebo (n = 13) each month for 3 years. The primary outcome was absolute and percentage change in total liver volume (TLV), which was measured by magnetic resonance imaging at baseline, after 3 years of treatment, and then 2 years after treatment ended. RESULTS Baseline characteristics were similar between groups. After 3 years, TLV decreased by 130.2 ± 133.2 mL in patients given octreotide LAR (7.8% ± 7.4%) (P = .003) but increased by 144.3 ± 316.8 mL (6.1% ± 14.1%) in patients given placebo. Change vs baseline differed significantly between groups (P = .004). Two years after treatment ended, TLV had decreased 14.4 ± 138.4 mL (0.8% ± 9.7%) from baseline in patients given octreotide LAR but increased by 224.4 ± 331.7 mL (11.0% ± 14.4%) in patients given placebo. Changes vs baseline still differed significantly between groups (P = .046). Decreases in TLV were similar in each sex; the change in TLV was greatest among subjects with larger baseline TLV. No patient withdrew because of side effects. CONCLUSIONS In a placebo-controlled study of patients with polycystic kidney and liver disease, 3 years of treatment with octreotide LAR significantly reduced liver volume; reductions were maintained for 2 years after treatment ended. Octreotide LAR was well-tolerated. ClinicalTrials.gov number: NCT02119052.
Collapse
Affiliation(s)
- Antonio Pisani
- Nephrology Unit, Department of Public Health, Federico II University, Naples, Italy
| | - Massimo Sabbatini
- Nephrology Unit, Department of Public Health, Federico II University, Naples, Italy
| | | | - Eleonora Riccio
- Nephrology Unit, Department of Public Health, Federico II University, Naples, Italy
| | - Nadia Rubis
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Clinical Research Centre for Rare Diseases "Aldo e Cele Daccò", Bergamo, Italy
| | - Anna Prinster
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Annalisa Perna
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Clinical Research Centre for Rare Diseases "Aldo e Cele Daccò", Bergamo, Italy
| | - Raffaele Liuzzi
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Letizia Spinelli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Michele Santangelo
- Department of Surgical Sciences and Nephrology, Federico II University, Naples, Italy
| | - Giuseppe Remuzzi
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Clinical Research Centre for Rare Diseases "Aldo e Cele Daccò", Bergamo, Italy; Nephrology and Dialysis Unit, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| | - Piero Ruggenenti
- IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Clinical Research Centre for Rare Diseases "Aldo e Cele Daccò", Bergamo, Italy; Nephrology and Dialysis Unit, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | |
Collapse
|
95
|
Maffezzoni F, Formenti AM, Mazziotti G, Frara S, Giustina A. Current and future medical treatments for patients with acromegaly. Expert Opin Pharmacother 2016; 17:1631-42. [PMID: 27352098 DOI: 10.1080/14656566.2016.1199687] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Acromegaly is a relatively rare condition of growth hormone (GH) excess associated with significant morbidity and, when left untreated, high mortality. Therapy for acromegaly is targeted at decreasing GH and insulin-like growth hormone 1 levels, ameliorating patients' symptoms and decreasing any local compressive effects of the pituitary adenoma. The therapeutic options for acromegaly include surgery, medical therapies (such as dopamine agonists, somatostatin receptor ligands and the GH receptor antagonist pegvisomant) and radiotherapy. However, despite all these treatments option, approximately 50% of patients are not adequately controlled. AREAS COVERED In this paper, the authors discuss: 1) efficacy and safety of current medical therapy 2) the efficacy and safety of the new multireceptor-targeted somatostatin ligand pasireotide 3) medical treatments currently under clinical investigation (oral octreotide, ITF2984, ATL1103), and 4) preliminary data on the use of new injectable and transdermal/transmucosal formulations of octreotide. EXPERT OPINION This expert opinion supports the need for new therapeutic agents and modalities for patients with acromegaly.
Collapse
Affiliation(s)
| | | | | | - Stefano Frara
- a Chair of Endocrinology , University of Brescia , Brescia , Italy
| | - Andrea Giustina
- a Chair of Endocrinology , University of Brescia , Brescia , Italy
| |
Collapse
|
96
|
Walls GV, Stevenson M, Soukup BS, Lines KE, Grossman AB, Schmid HA, Thakker RV. Pasireotide Therapy of Multiple Endocrine Neoplasia Type 1-Associated Neuroendocrine Tumors in Female Mice Deleted for an Men1 Allele Improves Survival and Reduces Tumor Progression. Endocrinology 2016; 157:1789-98. [PMID: 26990064 PMCID: PMC4870877 DOI: 10.1210/en.2015-1965] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pasireotide, a somatostatin analog, is reported to have anti-proliferative effects in neuroendocrine tumors (NETs). We therefore assessed the efficacy of pasireotide for treating pancreatic and pituitary NETs that develop in a mouse model of multiple endocrine neoplasia type 1 (MEN1). Men1(+/-) mice were treated from age 12 mo with 40 mg/kg pasireotide long-acting release formulation, or PBS, intramuscularly monthly for 9 mo. The Men1(+/-) mice had magnetic resonance imaging at 12 and 21 mo, and from 20 mo oral 5-bromo-2-deoxyuridine for 1 mo, to assess tumor development and proliferation, respectively. NETs were collected at age 21 mo, and proliferation and apoptosis assessed by immunohistochemistry and TUNEL assays, respectively. Pasireotide-treated Men1(+/-) mice had increased survival (pasireotide, 80.9% vs PBS, 65.2%; P < .05), with fewer mice developing pancreatic NETs (pasireotide, 86.9% vs PBS, 96.9%; P < .05) and smaller increases in pituitary NET volumes (pre-treated vs post-treated, 0.803 ± 0.058 mm(3) vs 2.872 ± 0.728 mm(3) [pasireotide] compared with 0.844 ± 0.066 mm(3) vs 8.847 ±1.948 mm(3) [PBS]; P < .01). In addition, pasireotide-treated mice had fewer pancreatic NETs compared with PBS-treated mice (2.36 ± 0.25 vs 3.72 ± 0.32, respectively; P < .001), with decreased proliferation in pancreatic NETs (pasireotide, 0.35 ± 0.03% vs PBS, 0.78 ± 0.08%; P < .0001) and pituitary NETs (pasireotide, 0.73 ±0.07% vs PBS, 1.81 ± 0.15%; P < .0001), but increased apoptosis in pancreatic NETs (pasireotide, 0.42 ± 0.05% vs PBS, 0.19 ± 0.03%; P < .001) and pituitary NETs (pasireotide, 14.75 ± 1.58% vs PBS, 2.35 ± 0.44%; P < .001). Thus, pasireotide increased survival and inhibited pancreatic and pituitary NET growth, thereby indicating its potential as an anti-proliferative and pro-apoptotic therapy.
Collapse
Affiliation(s)
- Gerard V Walls
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Mark Stevenson
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Benjamin S Soukup
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Kate E Lines
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Ashley B Grossman
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Herbert A Schmid
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Rajesh V Thakker
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| |
Collapse
|
97
|
Mazziotti G, Maffezzoni F, Giustina A. Vitamin D-binding protein: one more piece in the puzzle of acromegalic osteopathy? Endocrine 2016; 52:183-6. [PMID: 26899438 DOI: 10.1007/s12020-016-0890-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/02/2016] [Indexed: 12/21/2022]
Affiliation(s)
- G Mazziotti
- Endocrine and Metabolic Disease Unit, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
- Endocrine Unit, Department of Medicine, Carlo Poma Hospital, Mantua, Italy
| | - F Maffezzoni
- Endocrine and Metabolic Disease Unit, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
| | - A Giustina
- Endocrine and Metabolic Disease Unit, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| |
Collapse
|
98
|
Giustina A, Bevan JS, Bronstein MD, Casanueva FF, Chanson P, Petersenn S, Thanh XMT, Sert C, Houchard A, Guillemin I, Melmed S. SAGIT®: clinician-reported outcome instrument for managing acromegaly in clinical practice--development and results from a pilot study. Pituitary 2016; 19:39-49. [PMID: 26377024 PMCID: PMC4710645 DOI: 10.1007/s11102-015-0681-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE The SAGIT instrument is a comprehensive clinician-reported outcome instrument assessing key features of acromegaly: signs and symptoms, associated comorbidities; growth hormone levels; insulin-like growth factor-1 levels; and tumor profile. The SAGIT instrument has been designed to assist endocrinologists managing acromegaly in practice. Here, we report on pre-testing (to assess ease of understanding and acceptability) and a pilot study (to assess relevance, ease of use, and utility in real-life conditions) (NCT02231593). METHODS For pre-testing, 11 endocrinologists completed the SAGIT instrument using patient medical records and were also interviewed. They subsequently completed a PRAgmatic Content and face validity Test (PRAC-Test(©)) to report their experiences using SAGIT, and feedback was used to revise the instrument. In the pilot study, nine endocrinologists completed the SAGIT instrument in real-time with patients belonging to three different categories (stable/controlled, active/uncontrolled acromegaly, treatment-naïve), while four completed the instrument based on medical-record review. All participants then completed the PRAC-Test(©) and their feedback was used to update the instrument. RESULTS The SAGIT instrument was well accepted by endocrinologists, with most indicating that it was concise, practical, easy to understand, useful for assessing treatment response, and valuable as a component of the patient's medical record. The pilot study confirmed the instrument's acceptability, utility, and ease of use, and indicated its potential for distinguishing acromegaly clinical stages. CONCLUSIONS The SAGIT instrument is promising as a tool for use by endocrinologists in everyday practice to assess the status and evolution of disease in patients with acromegaly and to guide treatment decision-making.
Collapse
Affiliation(s)
- Andrea Giustina
- Chair of Endocrinology, University of Brescia, Brescia, Italy.
| | | | | | - Felipe F Casanueva
- Department of Medicine, Santiago de Compostela University - CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto Salud Carlos III, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Abstract
The currently available somatostatin receptor ligands (SRLs) and growth hormone (GH) antagonists are used to control levels of GH and insulin-like growth factor 1 (IGF-1) in patients with acromegaly. However, these therapies are limited by wide variations in efficacy, associated adverse effects and the need for frequent injections. A phase III trial of oral octreotide capsules demonstrated that this treatment can safely sustain suppressed levels of GH and IGF-1 and reduce the severity of symptoms in patients with acromegaly previously controlled by injectable SRL therapy, with the added benefit of no injection-site reactions. Phase I and phase II trials of the pan-selective SRL DG3173, the liquid crystal octreotide depot CAM2029 and an antisense oligonucleotide directed against the GH receptor have shown that these agents can be used to achieve biochemical suppression in acromegaly and have favourable safety profiles. This Review outlines the need for new therapeutic agents for patients with acromegaly, reviews clinical trial data of investigational agents and considers how these therapies might best be integrated into clinical practice.
Collapse
Affiliation(s)
- Shlomo Melmed
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 2015, Los Angeles, California 90048, USA
| |
Collapse
|
100
|
Claessen KMJA, Mazziotti G, Biermasz NR, Giustina A. Bone and Joint Disorders in Acromegaly. Neuroendocrinology 2016; 103:86-95. [PMID: 25633971 DOI: 10.1159/000375450] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/22/2015] [Indexed: 11/19/2022]
Abstract
Acromegaly is a chronic, progressive disease caused by a growth hormone (GH)-producing pituitary adenoma, resulting in elevated GH and insulin-like growth factor 1 concentrations. Following appropriate therapy (surgery, radiotherapy and/or medical treatment), many systemic GH-induced comorbid conditions improve considerably. Unfortunately, despite biochemical control, acromegaly patients suffer from a high prevalence of late manifestations of transient GH excess, significantly impairing their quality of life. In this overview article, we summarize the pathophysiology, diagnosis, clinical picture, disease course and management of skeletal complications of acromegaly, focusing on vertebral fractures and arthropathy.
Collapse
Affiliation(s)
- Kim M J A Claessen
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|